Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
N Engl J Med ; 378(13): 1189-1199, 2018 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-29601269

RESUMEN

BACKGROUND: Patients with acute myeloid leukemia (AML) often reach complete remission, but relapse rates remain high. Next-generation sequencing enables the detection of molecular minimal residual disease in virtually every patient, but its clinical value for the prediction of relapse has yet to be established. METHODS: We conducted a study involving patients 18 to 65 years of age who had newly diagnosed AML. Targeted next-generation sequencing was carried out at diagnosis and after induction therapy (during complete remission). End points were 4-year rates of relapse, relapse-free survival, and overall survival. RESULTS: At least one mutation was detected in 430 out of 482 patients (89.2%). Mutations persisted in 51.4% of those patients during complete remission and were present at various allele frequencies (range, 0.02 to 47%). The detection of persistent DTA mutations (i.e., mutations in DNMT3A, TET2, and ASXL1), which are often present in persons with age-related clonal hematopoiesis, was not correlated with an increased relapse rate. After the exclusion of persistent DTA mutations, the detection of molecular minimal residual disease was associated with a significantly higher relapse rate than no detection (55.4% vs. 31.9%; hazard ratio, 2.14; P<0.001), as well as with lower rates of relapse-free survival (36.6% vs. 58.1%; hazard ratio for relapse or death, 1.92; P<0.001) and overall survival (41.9% vs. 66.1%; hazard ratio for death, 2.06; P<0.001). Multivariate analysis confirmed that the persistence of non-DTA mutations during complete remission conferred significant independent prognostic value with respect to the rates of relapse (hazard ratio, 1.89; P<0.001), relapse-free survival (hazard ratio for relapse or death, 1.64; P=0.001), and overall survival (hazard ratio for death, 1.64; P=0.003). A comparison of sequencing with flow cytometry for the detection of residual disease showed that sequencing had significant additive prognostic value. CONCLUSIONS: Among patients with AML, the detection of molecular minimal residual disease during complete remission had significant independent prognostic value with respect to relapse and survival rates, but the detection of persistent mutations that are associated with clonal hematopoiesis did not have such prognostic value within a 4-year time frame. (Funded by the Queen Wilhelmina Fund Foundation of the Dutch Cancer Society and others.).


Asunto(s)
Análisis Mutacional de ADN , ADN de Neoplasias/análisis , Leucemia Mieloide Aguda/genética , Mutación , Neoplasia Residual/genética , Adolescente , Adulto , Anciano , Análisis Mutacional de ADN/métodos , Femenino , Citometría de Flujo , Hematopoyesis/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Leucemia Mieloide Aguda/mortalidad , Masculino , Persona de Mediana Edad , Neoplasia Residual/diagnóstico , Pronóstico , Modelos de Riesgos Proporcionales , Recurrencia , Inducción de Remisión , Análisis de Supervivencia , Adulto Joven
2.
Blood ; 119(24): 5824-31, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22490330

RESUMEN

The prevalence, the prognostic effect, and interaction with other molecular markers of DNMT3A mutations was studied in 415 patients with acute myeloid leukemia (AML) younger than 60 years. We show mutations in DNMT3A in 96 of 415 patients with newly diagnosed AML (23.1%). Univariate Cox regression analysis showed that patients with DNMT3A(mutant) AML show significantly worse overall survival (OS; P = .022; hazard ratio [HR], 1.38; 95% confidence interval [CI], 1.04-1.81), and relapse-free survival (RFS; P = .005; HR, 1.52; 95% CI, 1.13-2.05) than DNMT3A(wild-type) AMLs. In a multivariable analysis, DNMT3A mutations express independent unfavorable prognostic value for OS (P = .003; HR, 1.82; 95% CI, 1.2-2.7) and RFS (P < .001; HR, 2.2; 95% CI, 1.4-3.3). In a composite genotypic subset of cytogenetic intermediate-risk AML without FLT3-ITD and NPM1 mutations, this association is particularly evident (OS: P = .013; HR, 2.09; 95% CI, 1.16-3.77; RFS: P = .001; HR, 2.65; 95% CI, 1.48-4.89). The effect of DNMT3A mutations in human AML remains elusive, because DNMT3A(mutant) AMLs did not express a methylation or gene expression signature that discriminates them from patients with DNMT3A(wild-type) AML. We conclude that DNMT3A mutation status is an important factor to consider for risk stratification of patients with AML.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/genética , Proteínas Mutantes/metabolismo , Adolescente , Adulto , Biomarcadores de Tumor , Análisis por Conglomerados , Metilación de ADN/genética , ADN Metiltransferasa 3A , Femenino , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/terapia , Masculino , Persona de Mediana Edad , Análisis Multivariante , Mutación/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Nucleofosmina , Pronóstico , Recurrencia , Análisis de Supervivencia , Resultado del Tratamiento , Adulto Joven
3.
Nat Commun ; 15(1): 5693, 2024 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-38972954

RESUMEN

Leukemias with ambiguous lineage comprise several loosely defined entities, often without a clear mechanistic basis. Here, we extensively profile the epigenome and transcriptome of a subgroup of such leukemias with CpG Island Methylator Phenotype. These leukemias exhibit comparable hybrid myeloid/lymphoid epigenetic landscapes, yet heterogeneous genetic alterations, suggesting they are defined by their shared epigenetic profile rather than common genetic lesions. Gene expression enrichment reveals similarity with early T-cell precursor acute lymphoblastic leukemia and a lymphoid progenitor cell of origin. In line with this, integration of differential DNA methylation and gene expression shows widespread silencing of myeloid transcription factors. Moreover, binding sites for hematopoietic transcription factors, including CEBPA, SPI1 and LEF1, are uniquely inaccessible in these leukemias. Hypermethylation also results in loss of CTCF binding, accompanied by changes in chromatin interactions involving key transcription factors. In conclusion, epigenetic dysregulation, and not genetic lesions, explains the mixed phenotype of this group of leukemias with ambiguous lineage. The data collected here constitute a useful and comprehensive epigenomic reference for subsequent studies of acute myeloid leukemias, T-cell acute lymphoblastic leukemias and mixed-phenotype leukemias.


Asunto(s)
Islas de CpG , Metilación de ADN , Epigénesis Genética , Redes Reguladoras de Genes , Humanos , Metilación de ADN/genética , Islas de CpG/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Factor de Unión a CCCTC/metabolismo , Factor de Unión a CCCTC/genética , Regulación Leucémica de la Expresión Génica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cromatina/metabolismo , Cromatina/genética , Masculino , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Femenino , Hematopoyesis/genética , Niño , Transcriptoma , Proteínas Proto-Oncogénicas , Transactivadores
4.
Blood ; 117(1): 234-41, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-20855866

RESUMEN

DNA methylation patterns are frequently dysregulated in cancer, although little is known of the mechanisms through which specific gene sets become aberrantly methylated. The ecotropic viral integration site 1 (EVI1) locus encodes a DNA binding zinc-finger transcription factor that is aberrantly expressed in a subset of acute myeloid leukemia (AML) patients with poor outcome. We find that the promoter DNA methylation signature of EVI1 AML blast cells differs from those of normal CD34(+) bone marrow cells and other AMLs. This signature contained 294 differentially methylated genes, of which 238 (81%) were coordinately hypermethylated. An unbiased motif analysis revealed an overrepresentation of EVI1 binding sites among these aberrantly hypermethylated loci. EVI1 was capable of binding to these promoters in 2 different EVI1-expressing cell lines, whereas no binding was observed in an EVI1-negative cell line. Furthermore, EVI1 was observed to interact with DNA methyl transferases 3A and 3B. Among the EVI1 AML cases, 2 subgroups were recognized, of which 1 contained AMLs with many more methylated genes, which was associated with significantly higher levels of EVI1 than in the cases of the other subgroup. Our data point to a role for EVI1 in directing aberrant promoter DNA methylation patterning in EVI1 AMLs.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/genética , Leucemia Mieloide Aguda/genética , Regiones Promotoras Genéticas/genética , Proto-Oncogenes/genética , Factores de Transcripción/genética , Adolescente , Adulto , Anciano , Western Blotting , Inmunoprecipitación de Cromatina , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , ADN de Neoplasias/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Proteína del Locus del Complejo MDS1 y EV11 , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Factores de Transcripción/metabolismo , Adulto Joven , ADN Metiltransferasa 3B
5.
Haematologica ; 97(3): 388-92, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22058207

RESUMEN

Somatic mutations in the additional sex comb-like 1 (ASXL1) gene have been described in various types of myeloid malignancies, including acute myeloid leukemia. Analysis of novel markers, such as ASXL1 mutations, in independent clinical trials is indispensable before considering them for clinical decision-making. We analyzed 882 well-characterized acute myeloid leukemia cases to determine the prevalence and prognostic impact of ASXL1 exon12 mutations. Truncating ASXL1 mutations were present in 46 cases (5.3%). ASXL1 mutations were inversely associated with FLT3 internal tandem duplications and mutually exclusive with NPM1 mutations. ASXL1 mutations were an unfavorable prognostic factor as regards survival (median overall survival 15.9 months vs. 22.3 months; P=0.019), with a significantly lower complete response rate (61% vs. 79.6%; P=0.004). In multivariate analyses, ASXL1 mutations were independently associated with inferior poor overall survival (HR 1.52, P=0.032). In conclusion, ASXL1 mutations are common mutations in acute myeloid leukemia and indicate a poor therapy outcome.


Asunto(s)
Leucemia Mieloide Aguda/genética , Mutación , Proteínas Represoras/genética , Adolescente , Adulto , Anciano , Femenino , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/epidemiología , Masculino , Persona de Mediana Edad , Nucleofosmina , Prevalencia , Pronóstico , Análisis de Supervivencia , Adulto Joven
6.
Blood ; 113(13): 3088-91, 2009 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-19171880

RESUMEN

Mutations in CCAAT/enhancer binding protein alpha (CEBPA) are seen in 5% to 14% of acute myeloid leukemia (AML) and have been associated with a favorable clinical outcome. Most AMLs with CEBPA mutations simultaneously carry 2 mutations (CEBPA(double-mut)), usually biallelic, whereas single heterozygous mutations (CEBPA(single-mut)) are less frequently seen. Using denaturing high-performance liquid chromatography and nucleotide sequencing, we identified among a cohort of 598 newly diagnosed AMLs a subset of 41 CEBPA mutant cases (28 CEBPA(double-mut) and 13 CEBPA(single-mut) cases). CEBPA(double-mut) associated with a unique gene expression profile as well as favorable overall and event-free survival, retained in multivariable analysis that included cytogenetic risk, FLT3-ITD and NPM1 mutation, white blood cell count, and age. In contrast, CEBPA(single-mut) AMLs did not express a discriminating signature and could not be distinguished from wild-type cases as regards clinical outcome. These results demonstrate significant underlying heterogeneity within CEBPA mutation-positive AML with prognostic relevance.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Perfilación de la Expresión Génica , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Mutación Missense , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Regulación Leucémica de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Leucemia Mieloide Aguda/clasificación , Leucemia Mieloide Aguda/mortalidad , Mutación Missense/fisiología , Nucleofosmina , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Análisis de Supervivencia
7.
Blood ; 113(12): 2795-804, 2009 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-19168792

RESUMEN

Acute myeloid leukemia is a heterogeneous disease from the molecular and biologic standpoints, and even patients with a specific gene expression profile may present clinical and molecular heterogeneity. We studied the epigenetic profiles of a cohort of patients who shared a common gene expression profile but differed in that only half of them harbored mutations of the CEBPA locus, whereas the rest presented with silencing of this gene and coexpression of certain T-cell markers. DNA methylation studies revealed that these 2 groups of patients could be readily segregated in an unsupervised fashion based on their DNA methylation profiles alone. Furthermore, CEBPA silencing was associated with the presence of an aberrant DNA hypermethylation signature, which was not present in the CEBPA mutant group. This aberrant hypermethylation occurred more frequently at sites within CpG islands. CEBPA-silenced leukemias also displayed marked hypermethylation compared with normal CD34(+) hematopoietic cells, whereas CEBPA mutant cases showed only mild changes in DNA methylation compared with these normal progenitors. Biologically, CEBPA-silenced leukemias presented with a decreased response to myeloid growth factors in vitro.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Metilación de ADN , ADN de Neoplasias/química , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Leucemia Mieloide Aguda/genética , Adolescente , Adulto , Estudios de Cohortes , Islas de CpG/genética , ADN de Neoplasias/genética , Resistencia a Antineoplásicos/genética , Regulación Leucémica de la Expresión Génica , Silenciador del Gen , Factor Estimulante de Colonias de Granulocitos/farmacología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Células Madre Hematopoyéticas/metabolismo , Humanos , Interleucina-3/farmacología , Leucemia Mieloide Aguda/clasificación , Leucemia Mieloide Aguda/patología , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/clasificación , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Células Tumorales Cultivadas/citología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
8.
Blood ; 114(14): 3001-7, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19666867

RESUMEN

Minimally differentiated acute myeloid leukemia (AML-M0) is defined by immature morphology and expression of early hematologic markers. By gene expression profiling (GEP) and subsequent unsupervised analysis of 35 AML-M0 samples and 253 previously reported AML cases, we demonstrate that AML-M0 cases express a unique signature that is largely separated from other molecular subtypes. Hematologic transcription regulators such as CEBPA, CEBPD, and ETV6, and the differentiation associated gene MPO appeared strongly down-regulated, in line with the primitive state of this leukemia. AML-M0 frequently carries loss-of-function RUNX1 mutation. Unsupervised analyses revealed a subdivision between AML-M0 cases with and without RUNX1 mutations. RUNX1 mutant AML-M0 samples showed a distinct up-regulation of B cell-related genes such as members of the B-cell receptor complex, transcription regulators RUNX3, ETS2, IRF8, or PRDM1, and major histocompatibility complex class II genes. Importantly, prediction with high accuracy of the AML-M0 subtype and prediction of patients carrying RUNX1 mutation within this subtype were possible based on the expression level of only a few transcripts. We propose that RUNX1 mutations in this AML subgroup cause lineage infidelity, leading to aberrant coexpression of myeloid and B-lymphoid genes. Furthermore, our results imply that AML-M0, although originally determined by morphology, constitutes a leukemia subgroup.


Asunto(s)
Biomarcadores de Tumor/genética , Diferenciación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/clasificación , Leucemia Mieloide Aguda/genética , Mutación/genética , Linfocitos B/patología , Redes Reguladoras de Genes , Humanos , Leucemia Mieloide Aguda/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Regulación hacia Arriba
9.
Nat Commun ; 12(1): 5679, 2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34584081

RESUMEN

Chromosomal rearrangements are a frequent cause of oncogene deregulation in human malignancies. Overexpression of EVI1 is found in a subgroup of acute myeloid leukemia (AML) with 3q26 chromosomal rearrangements, which is often therapy resistant. In AMLs harboring a t(3;8)(q26;q24), we observed the translocation of a MYC super-enhancer (MYC SE) to the EVI1 locus. We generated an in vitro model mimicking a patient-based t(3;8)(q26;q24) using CRISPR-Cas9 technology and demonstrated hyperactivation of EVI1 by the hijacked MYC SE. This MYC SE contains multiple enhancer modules, of which only one recruits transcription factors active in early hematopoiesis. This enhancer module is critical for EVI1 overexpression as well as enhancer-promoter interaction. Multiple CTCF binding regions in the MYC SE facilitate this enhancer-promoter interaction, which also involves a CTCF binding site upstream of the EVI1 promoter. We hypothesize that this CTCF site acts as an enhancer-docking site in t(3;8) AML. Genomic analyses of other 3q26-rearranged AML patient cells point to a common mechanism by which EVI1 uses this docking site to hijack enhancers active in early hematopoiesis.


Asunto(s)
Factor de Unión a CCCTC/genética , Elementos de Facilitación Genéticos/genética , Leucemia Mieloide/genética , Proteína del Locus del Complejo MDS1 y EV11/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proto-Oncogenes/genética , Enfermedad Aguda , Factor de Unión a CCCTC/metabolismo , Cromosomas Humanos Par 3/genética , Cromosomas Humanos Par 8/genética , Regulación Leucémica de la Expresión Génica , Reordenamiento Génico , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Hibridación Fluorescente in Situ/métodos , Células K562 , Cariotipificación , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Regiones Promotoras Genéticas/genética , Unión Proteica , Translocación Genética
10.
Cancer Discov ; 11(11): 2868-2883, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33980539

RESUMEN

In acute myeloid leukemia (AML) with inv(3)(q21;q26) or t(3;3)(q21;q26), a translocated GATA2 enhancer drives oncogenic expression of EVI1. We generated an EVI1-GFP AML model and applied an unbiased CRISPR/Cas9 enhancer scan to uncover sequence motifs essential for EVI1 transcription. Using this approach, we pinpointed a single regulatory element in the translocated GATA2 enhancer that is critically required for aberrant EVI1 expression. This element contained a DNA-binding motif for the transcription factor MYB, which specifically occupied this site at the translocated allele and was dispensable for GATA2 expression. MYB knockout as well as peptidomimetic blockade of CBP/p300-dependent MYB functions resulted in downregulation of EVI1 but not of GATA2. Targeting MYB or mutating its DNA-binding motif within the GATA2 enhancer resulted in myeloid differentiation and cell death, suggesting that interference with MYB-driven EVI1 transcription provides a potential entry point for therapy of inv(3)/t(3;3) AMLs. SIGNIFICANCE: We show a novel paradigm in which chromosomal aberrations reveal critical regulatory elements that are nonfunctional at their endogenous locus. This knowledge provides a rationale to develop new compounds to selectively interfere with oncogenic enhancer activity.This article is highlighted in the In This Issue feature, p. 2659.


Asunto(s)
Elementos de Facilitación Genéticos , Genes myb , Leucemia Mieloide Aguda , Translocación Genética , Factor de Transcripción GATA2 , Humanos , Leucemia Mieloide Aguda/genética , Proteína del Locus del Complejo MDS1 y EV11 , Oncogenes
11.
Cell Rep Med ; 1(5): 100074, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-33205068

RESUMEN

Severe congenital neutropenia (SCN) patients treated with CSF3/G-CSF to alleviate neutropenia frequently develop acute myeloid leukemia (AML). A common pattern of leukemic transformation involves the appearance of hematopoietic clones with CSF3 receptor (CSF3R) mutations in the neutropenic phase, followed by mutations in RUNX1 before AML becomes overt. To investigate how the combination of CSF3 therapy and CSF3R and RUNX1 mutations contributes to AML development, we make use of mouse models, SCN-derived induced pluripotent stem cells (iPSCs), and SCN and SCN-AML patient samples. CSF3 provokes a hyper-proliferative state in CSF3R/RUNX1 mutant hematopoietic progenitors but does not cause overt AML. Intriguingly, an additional acquired driver mutation in Cxxc4 causes elevated CXXC4 and reduced TET2 protein levels in murine AML samples. Expression of multiple pro-inflammatory pathways is elevated in mouse AML and human SCN-AML, suggesting that inflammation driven by downregulation of TET2 activity is a critical step in the malignant transformation of SCN.


Asunto(s)
Transformación Celular Neoplásica/genética , Síndromes Congénitos de Insuficiencia de la Médula Ósea/genética , Síndromes Congénitos de Insuficiencia de la Médula Ósea/patología , Proteínas de Unión al ADN/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Mutación/genética , Neutropenia/congénito , Factores de Transcripción/genética , Animales , Línea Celular , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células HEK293 , Humanos , Inflamación/genética , Inflamación/patología , Células K562 , Ratones , Neutropenia/genética , Neutropenia/patología , Transducción de Señal/genética
14.
Cancer Cell ; 17(1): 13-27, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20060365

RESUMEN

We hypothesized that DNA methylation distributes into specific patterns in cancer cells, which reflect critical biological differences. We therefore examined the methylation profiles of 344 patients with acute myeloid leukemia (AML). Clustering of these patients by methylation data segregated patients into 16 groups. Five of these groups defined new AML subtypes that shared no other known feature. In addition, DNA methylation profiles segregated patients with CEBPA aberrations from other subtypes of leukemia, defined four epigenetically distinct forms of AML with NPM1 mutations, and showed that established AML1-ETO, CBFb-MYH11, and PML-RARA leukemia entities are associated with specific methylation profiles. We report a 15 gene methylation classifier predictive of overall survival in an independent patient cohort (p < 0.001, adjusted for known covariates).


Asunto(s)
Biomarcadores de Tumor/genética , Metilación de ADN/genética , Epigénesis Genética/genética , Leucemia Mieloide Aguda/clasificación , Leucemia Mieloide Aguda/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/mortalidad , Masculino , Persona de Mediana Edad , Nucleofosmina , Pronóstico
15.
Blood ; 111(3): 1634-43, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18025157

RESUMEN

PML-RARalpha is the causative oncogene in 5% to 10% of the cases of acute myeloid leukemia. At physiological concentrations of retinoic acid, PML-RARalpha silences RARalpha target genes, blocking differentiation of the cells. At high concentrations of ligand, it (re)activates the transcription of target genes, forcing terminal differentiation. The study of RARalpha target genes that mediate this differentiation has identified several genes that are important for proliferation and differentiation control in normal and malignant hematopoietic cells. In this paper, we show that the PML-RARalpha fusion protein not only interferes with the transcription of regular RARalpha target genes. We show that the ID1 and ID2 promoters are activated by PML-RARalpha but, unexpectedly, not by wild-type RARalpha/RXR. Our data support a model in which the PML-RARalpha fusion protein regulates a novel class of target genes by interaction with the Sp1 and NF-Y transcription factors, without directly binding to the DNA, defining a gain-of-function for the oncoprotein.


Asunto(s)
ADN/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Activación Transcripcional/genética , Factor de Unión a CCAAT/genética , Factor de Unión a CCAAT/metabolismo , Línea Celular Tumoral , ADN/metabolismo , Humanos , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/patología , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Unión Proteica , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Activación Transcripcional/efectos de los fármacos , Tretinoina/farmacología , Regulación hacia Arriba
16.
Blood ; 110(10): 3706-14, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17671232

RESUMEN

Gene expression profiling of acute myeloid leukemia (AML) allows the discovery of previously unrecognized molecular entities. Here, we identified a specific subgroup of AML, defined by an expression profile resembling that of AMLs with mutations in the myeloid transcription factor CCAAT/enhancer-binding protein alpha (C/EBPalpha), while lacking such mutations. We found that in these leukemias, the CEBPA gene was silenced, which was associated with frequent promoter hypermethylation. The leukemias phenotypically showed aberrant expression of T-cell genes, of which CD7 was most consistent. We identified 2 mechanisms that may contribute to this phenotype. First, absence of Cebpa led to up-regulation of specific T-cell transcripts (ie, Cd7 and Lck) in hematopoietic stem cells isolated from conditional Cebpa knockout mice. Second, the enhanced expression of TRIB2, which we identify here as a direct target of the T-cell commitment factor NOTCH1, suggested aberrantly activated Notch signaling. Putatively activating NOTCH1 mutations were found in several specimens of the newly identified subgroup, while a large set of control AMLs was mutation negative. A gene expression prediction signature allowed the detection of similar cases of leukemia in independent series of AML.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Silenciador del Gen , Leucemia Mieloide Aguda/genética , Leucemia-Linfoma de Células T del Adulto/genética , Receptor Notch1/genética , Animales , Células Cultivadas , Análisis por Conglomerados , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos
17.
Blood ; 106(13): 4114-23, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16118314

RESUMEN

Protein ubiquitination plays important roles in a variety of basic cellular processes. Proteins are ubiquitinated by E2-E3 ubiquitin ligase complexes. Depending on the type of ubiquitin chain conjugated, proteins are either targeted for degradation by the proteasome or their activity is specifically altered. We describe a novel conserved nuclear protein, Triad1 (2 RING [really interesting new gene] fingers and DRIL [double RING finger linked] 1), which is strongly induced during myeloid differentiation. Triad1 contains a TRIAD motif that harbors 2 RING finger structures. Triad1 binds the E2 ubiquitin-conjugating enzyme UbcH7 as well as ubiquitinated proteins and supports the formation of ubiquitin chains that are recognized by the proteasome. The biologic function of Triad1 in myelopoiesis was studied by performing granulocyte-macrophage colony-forming unit (CFU-GM) assays using retrovirally transduced primary murine bone marrow cells. Triad1 severely inhibited myeloid colony formation. In contrast, 2 Triad1 RING finger point mutants that failed to bind UbcH7 did not affect colony formation. Moreover, proteasome inhibition counteracted the inhibition of colony formation exerted by wild-type Triad1. In liquid cultures, Triad1 did not influence differentiation but strongly inhibited proliferation resulting in a G0/G1 accumulation. We conclude that proteasomal degradation of proteins that are ubiquitinated by Triad1 affects the clonogenic growth of primary myeloid progenitor cells.


Asunto(s)
Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Diferenciación Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular , Células Cultivadas , Granulocitos/citología , Granulocitos/metabolismo , Humanos , Ratones , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Células Tumorales Cultivadas , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA